Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
J Periodontol ; 2023 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-37885337

RESUMO

BACKGROUND: Stannous fluoride dentifrice is well established for its beneficial clinical effects. In this study, we evaluated the effects of stannous fluoride on inflammation and oral microbiome. METHODS: In this randomized, parallel-arm, double-blind, controlled clinical trial, we compared clinical resolution of experimental gingivitis by evaluating bleeding on probing, gingival index, and plaque index between stannous fluoride stabilized with zinc phosphate (test) and sodium fluoride (control) dentifrices. Further, these groups were compared for oral neutrophil counts, systemic priming of neutrophils, gingival crevicular fluid (GCF) expression of inflammatory markers, and the oral microbiome. RESULTS: We found significant reduction in bleeding on probing in the test group compared to the control group in experimental gingivitis when participants used the test dentifrice prior to induction of experimental gingivitis. The test group also showed significant reductions in GCF levels of inflammatory markers (matrix metalloproteinase 8 [MMP8], receptor activator of nuclear factor kappa-Β ligand [RANKL]), oral polymorphonuclear neutrophil (PMN) counts, and systemic neutrophil priming (CD11b expression) during experimental gingivitis. Further, significant reductions in the gram-negative genera Porphyromonas, Tannerella, and Treponema were noted in the test group. CONCLUSION: The stannous fluoride stabilized with zinc phosphate dentifrice formulation demonstrated clinical reduction in gingival inflammation and a beneficial effect on microbiome and immune markers. This intervention should be explored as a preventive aid in the progression of plaque-induced gingivitis to periodontitis.

2.
Proc Natl Acad Sci U S A ; 120(41): e2306020120, 2023 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-37782795

RESUMO

Variation in human immune response to the same bacterial or viral pathogen is well established in the literature. Variation in immune response to microbial challenge has also been observed within the human oral cavity. Our recent study focused on characterizing observed variations in microbially induced gingival inflammation-resulting in three distinct clinical Inflammatory Responder Types (IRTs): High-IRT, Low-IRT, and Slow-IRT. Here, we applied a high-resolution temporal multiomic analysis during microbially induced inflammation in order to characterize the effects of localized oral inflammation on distant healthy tissues in young healthy adults. Our results highlight a nonlocalized subclinical effect with alterations in proinflammatory host mediators and an ecological shift toward dysbiosis within the subgingival microbiome in an IRT-dependent manner-despite maintained oral hygiene. Our results provide mechanistic insight into how healthy tissues within humans are influenced by distant localized inflammation and may ultimately become susceptible to disease.


Assuntos
Gengivite , Microbiota , Adulto , Humanos , Gengivite/microbiologia , Inflamação , Bactérias
3.
Front Oral Health ; 3: 911420, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35677024

RESUMO

The oral microbiome, with a unique emphasis on Porphyromonas gingivalis has been associated with a constellation of inflammatory diseases such as cardiovascular disease, rheumatoid arthritis, Alzheimer's disease, type II diabetes, and non-alcoholic associated fatty liver disease. Periodontal disease has also been shown to induce "leaky gut" leading to metabolic endotoxemia. Several recent studies investigating the habitants of the blood microbiome have found the majority of species appear to be derived from oral and skin bacterial communities in otherwise healthy individuals. Many of the same pathologies associated with perturbations of oral health, such as cardiovascular disease, show alterations to the composition of the blood microbiome as well as circulating neutrophil phenotypes. Gingival inflammation is associated with activated blood neutrophil phenotypes that can exacerbate a distal inflammatory insult which may explain the connection between oral and systemic inflammatory conditions. While in the oral cavity, neutrophils encounter oral microbes that are adept in manipulating neutrophil activity which can re-enter the vasculature thereafter. Endotoxin from oral microbes can differ significantly depending on bacterial community and state of oral health to alter cellular LPS tolerance mechanisms which may contribute to the primed neutrophil phenotype seen in periodontitis and provide a mechanism by which the oral-microbes can affect systemic health outcomes. This review synthesizes the studies between inflammatory diseases and oral health with emphasis on microbiome and corresponding lipopolysaccharides in immune tolerance and activation.

4.
J Periodontol ; 93(12): 1940-1950, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35100435

RESUMO

BACKGROUND: The gingival epithelium protects periodontal tissues and the alveolar bone by maintaining a steady state of regulated inflammatory surveillance, also known as healthy homeostasis. Accordingly, the repertoire of receptors present within the gingival epithelium showcases its ability to recognize microbial colonization and contribute to bacterial sensing. Macrophage migration inhibitory factor (MIF) is one of many cytokines that are expressed in this protective state and is involved in neutrophil regulation. However, its role in the maintenance of healthy gingival tissue has not been described. METHODS: Gingival tissues from wild-type (WT) and Mif knock-out (KO) mice were stained for neutrophils and three key neutrophil chemoattractants: MIF, Gro-α/CXCL1, and Gro-ß/CXCL2 in the junctional epithelium (JE). In addition, gene silencing studies were performed using gingival epithelial cells (GECs) to examine the role of MIF on transcription of key bacterial recognition receptors Toll-like receptors (TLR)-1, -2, -4, -6, -9 and interleukin-1 receptors (IL-1R1 and IL-1R2) in response to oral bacterial stimulation. RESULTS: WT murine gingival tissues demonstrated high expression of MIF in the JE. In Mif KO mice, despite the significant reduction of Gro-α/CXCL1 and Gro-ß/CXCL2, there was a slight increase in neutrophils. Gene silencing experiments showed that MIF down-regulated the mRNA expression of TLR4, IL-1R1, and IL-1R2 in GEC, in addition to decreasing secreted IL-8/CXCL8 in response to bacteria. CONCLUSIONS: MIF regulates the expression of TLR4, IL-1Rs, and IL-8/CXCL8, components that are all involved in maintaining oral health. Our data demonstrate that MIF is a significant contributor to the maintenance of healthy oral homeostasis.


Assuntos
Células Epiteliais , Imunidade Inata , Fatores Inibidores da Migração de Macrófagos , Animais , Camundongos , Interleucina-8 , Camundongos Knockout , Receptores Tipo II de Interleucina-1 , Receptor 4 Toll-Like , Gengiva/citologia
5.
JCI Insight ; 7(4)2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35077397

RESUMO

Commensal microbes critically regulate skeletal homeostasis, yet the impact of specific microbiota communities on osteoimmune response mechanisms is unknown. To discern osteoimmunomodulatory effects imparted by the commensal oral microbiota that are distinct from the systemic microbiota, osteoimmunology studies were performed in both alveolar bone and nonoral skeletal sites of specific pathogen-free (SPF) versus germ-free (GF) mice and SPF mice subjected to saline versus chlorhexidine oral rinses. SPF versus GF mice had reduced cortical/trabecular bone and an enhanced pro-osteoclastic phenotype in alveolar bone. TLR signaling and Th17 cells that have known pro-osteoclastic actions were increased in alveolar BM, but not long BM, of SPF versus GF mice. MHC II antigen presentation genes and activated DCs and CD4+ T cells were elevated in alveolar BM, but not long BM, of SPF versus GF mice. These findings were substantiated by in vitro allostimulation studies demonstrating increased activated DCs derived from alveolar BM, but not long BM, of SPF versus GF mice. Chlorhexidine antiseptic rinse depleted the oral, but not gut, bacteriome in SPF mice. Findings from saline- versus chlorhexidine-treated SPF mice corroborated outcomes from SPF versus GF mice, which reveals that the commensal oral microbiota imparts osteoimmunomodulatory effects separate from the systemic microbiome.


Assuntos
Microbioma Gastrointestinal/imunologia , Vida Livre de Germes/imunologia , Boca/microbiologia , Osteoclastos/imunologia , Organismos Livres de Patógenos Específicos/imunologia , Animais , Homeostase/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais
6.
Cell Host Microbe ; 29(10): 1589-1598.e6, 2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34536346

RESUMO

Colorectal cancer is a major health concern worldwide. Growing evidence for the role of the gut microbiota in the initiation of CRC has sparked interest in approaches that target these microorganisms. However, little is known about the composition and role of the microbiota associated with precancerous polyps. Here, we found distinct microbial signatures between patients with and without polyps and between polyp subtypes using sequencing and culturing techniques. We found a correlation between Bacteroides fragilis recovered and the level of inflammatory cytokines in the mucosa adjacent to the polyp. Additional analysis revealed that B. fragilis from patients with polyps are bft-negative, activate NF-κB through Toll-like receptor 4, induce a pro-inflammatory response, and are enriched in genes associated with LPS biosynthesis. This study provides fundamental insight into the microbial microenvironment of the pre-neoplastic polyp by highlighting strain-specific genomic and proteomic differences, as well as more broad compositional differences in the microbiome.


Assuntos
Bacteroides fragilis/genética , Bacteroides fragilis/isolamento & purificação , Neoplasias Colorretais/microbiologia , Mucosa Intestinal/microbiologia , Idoso , Bacteroides fragilis/classificação , Bacteroides fragilis/fisiologia , Pólipos do Colo/imunologia , Pólipos do Colo/microbiologia , Pólipos do Colo/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Citocinas/genética , Citocinas/imunologia , Feminino , Microbioma Gastrointestinal , Genoma Bacteriano , Genômica , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Filogenia , Simbiose
7.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34193520

RESUMO

Oral commensal bacteria actively participate with gingival tissue to maintain healthy neutrophil surveillance and normal tissue and bone turnover processes. Disruption of this homeostatic host-bacteria relationship occurs during experimental gingivitis studies where it has been clearly established that increases in the bacterial burden increase gingival inflammation. Here, we show that experimental gingivitis resulted in three unique clinical inflammatory phenotypes (high, low, and slow) and reveal that interleukin-1ß, a reported major gingivitis-associated inflammatory mediator, was not associated with clinical gingival inflammation in the slow response group. In addition, significantly higher levels of Streptococcus spp. were also unique to this group. The low clinical response group was characterized by low concentrations of host mediators, despite similar bacterial accumulation and compositional characteristics as the high clinical response group. Neutrophil and bone activation modulators were down-regulated in all response groups, revealing novel tissue and bone protective responses during gingival inflammation. These alterations in chemokine and microbial composition responses during experimental gingivitis reveal a previously uncharacterized variation in the human host response to a disruption in gingival homeostasis. Understanding this human variation in gingival inflammation may facilitate the identification of periodontitis-susceptible individuals. Overall, this study underscores the variability in host responses in the human population arising from variations in host immune profiles (low responders) and microbial community maturation (slow responders) that may impact clinical outcomes in terms of destructive inflammation.


Assuntos
Gengiva/patologia , Inflamação/patologia , Adolescente , Adulto , Osso e Ossos/patologia , Quimiocinas/metabolismo , Gengiva/microbiologia , Gengivite/microbiologia , Gengivite/patologia , Homeostase , Humanos , Filogenia , Fatores de Tempo , Adulto Jovem
8.
J Endod ; 47(8): 1265-1271, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34015415

RESUMO

INTRODUCTION: We verified the association between selected bacterial profiles and levels of cytokines, chemokines, and the expression of signs and symptoms of primary endodontic infection with apical periodontitis. METHODS: Samples were collected from 21 root canals, and macrophages were stimulated for 24 hours. Tumor necrosis factor alpha (TNF-α), interleukin (IL)-6, IL-10, IL-12p70, interferon gamma, and chemokine (C-C motif) ligand 2 (CCL2) were measured using cytometric bead array. We investigated the overlapping networks between cytokines and chemokines with regression analysis. Checkerboard DNA-DNA hybridization was used to assess 40 target bacteria species. Using factor analysis, bacterial species aggregated in 2 factors. The association of bacteria species-based factors on cytokine and chemokine levels and clinical features was estimated with regression analysis. RESULTS: A negative relationship between IL-10 (anti-inflammatory cytokine) and CCL2, TNF-α, and IFN-γ (proinflammatory cytokines) (all P < .05) was observed. CCL2 was positively correlated with TNF-α (P < .01). Thirty-eight bacteria species were detected in primary endodontic infection with apical periodontitis. The first bacteria species-based factor was associated with the size of the radiolucent area (coefficient = 15.42) and tenderness to percussion/pain on palpation (coefficient = 20.79). The second factor was associated with CCL2 levels (coefficient = 1.28). CONCLUSIONS: Different bacterial profiles can be differentially related to the expression of inflammatory proteins and the experience of clinical features.


Assuntos
Citocinas , Periodontite Periapical , Bactérias , Quimiocina CCL2 , Humanos , Interleucina-6 , Fator de Necrose Tumoral alfa
9.
Periodontol 2000 ; 86(1): 188-200, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33690934

RESUMO

Years of coevolution with resident microbes has made them an essential component of health. Yet, little is known about oral commensal bacteria's contribution to and role in the maintenance of oral health and homeostasis. Commensal bacteria are speculated to play a host protective role in the maintenance of health. In this review, we describe and provide examples of the coordinate regulation that occurs between oral commensal bacteria and the host innate immune response to modulate and maintain oral homeostasis.


Assuntos
Imunidade Inata , Microbiota , Epitélio , Homeostase , Humanos , Periodonto
10.
Periodontol 2000 ; 86(1): 8-13, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33690952

RESUMO

The central theme of this volume of Periodontology 2000 is that the microbial dental plaque biofilm, specifically the subgingival dental plaque biofilm, mimics a human tissue in both structure and function. As a basis for this assertion we use the definition of a tissue as an aggregate of similar cells and cell products forming a defined structure with a specific function, in a multicellular organism. Accordingly, we propose that the dental plaque biofilm represents an acquired human tissue largely of bacterial origin that maintains the health of gingival tissue. Furthermore, we acknowledge that disease can be defined as a deviation from the normal structure or an interruption to the function of any body part, organ, or system, and that is manifested by a characteristic set of symptoms and signs whose etiology, pathology, and prognosis may be known or unknown. Therefore, in this volume we present the concept that periodontitis is a disruption of the normal function of the healthy subgingival plaque biofilm with concomitant disruption to its functional properties in relation to innate defense surveillance and tissue maintenance, leading to excessive, deregulated inflammation and tissue destruction.


Assuntos
Placa Dentária , Periodontite , Biofilmes , Gengiva , Humanos
12.
Front Oral Health ; 2: 689475, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35048035

RESUMO

Aim: Clinically healthy gingival tissue is maintained through controlled regulation of host defense mechanisms against plaque biofilm overgrowth. One key component is the transit of neutrophils from the vasculature into gingival tissue where the expression of different neutrophil chemokines are tightly regulated. This cross-sectional study examines the inter-individual variability in chemokine profiles within gingival crevicular fluid (GCF) in relation to the subgingival bacterial community in a state of gingival health. Methods: Gingival crevicular fluid and subgingival plaque samples were collected from mesiobuccal surfaces of all six Ramfjord teeth of 20 systemically healthy individuals (14.55 ± 1.67 years). A multiplex immunoassay was carried out to quantify the expression of 40 different chemokines in the healthy gingival tissue. Neutrophils were assessed indirectly by myeloperoxidase (MPO) in GCF using traditional ELISA. Characterization of healthy subgingival plaque was conducted with the Illumina Miseq targeting the 16S rRNA gene. Results: In health, there are distinct variations within individual gingival crevicular fluid chemokine expression profiles, as well as in the concentration of neutrophils, that divided the participants into high or low chemokine expressing groups. Specifically, key differences were identified within MIF (2683.54 ± 985.82 pg per 30-s sample), IL-8/CXCL8 (170.98 ± 176.96 pg per 30-s sample), Gro-α/CXCL1 (160.42 ± 94.21 pg per 30-s sample), ENA-78/CXCL5 (137.76 ± 76.02 pg per 30-s sample), IL-1ß (51.39 ± 37.23 pg per 30-s sample), TNF-α (1.76 ± 1.79 pg per 30-s sample), and IFN-γ (0.92 ± 0.54 pg per 30-s sample). Of these identified chemokines, the highest correlation was associated between IL-8/CXCL8 and neutrophils (r = 0.54, p = 0.014). Furthermore, species characterization of healthy subgingival plaque revealed significant inter-individual variability that identified two unique groups unrelated to the previously identified chemokine groups. Conclusion: The lack of concordance between the microbial composition and chemokine profile during health may be a reflection of the unique microbial composition of each individual coupled with variations within their host response, emphasizing the vast complexity of the defense mechanisms in place to maintain gingival health.

13.
Infect Immun ; 89(2)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33257533

RESUMO

The majority of Gram-negative bacteria elicit a potent immune response via recognition of lipid A expressed on the outer bacterial membrane by the host immune receptor Toll-like receptor 4 (TLR4). However, some Gram-negative bacteria evade detection by TLR4 or alter the outcome of TLR4 signaling by modification of lipid A species. Although the role of lipid A modifications on host innate immunity has been examined in some detail, it is currently unclear how lipid A remodeling influences host adaptive immunity. One prototypic Gram-negative bacterium that modifies its lipid A structure is Porphyromonas gingivalis, an anaerobic pathobiont that colonizes the human periodontium and induces chronic low-grade inflammation that is associated with periodontal disease as well as a number of systemic inflammatory disorders. P. gingivalis produces dephosphorylated and deacylated lipid A structures displaying altered activities at TLR4. Here, we explored the functional role of P. gingivalis lipid A modifications on TLR4-dependent innate and adaptive immune responses in mouse bone marrow-derived dendritic cells (BMDCs). We discovered that lipid A 4'-phosphate removal is required for P. gingivalis to evade BMDC-dependent proinflammatory cytokine responses and markedly limits the bacterium's capacity to induce beta interferon (IFN-ß) production. In addition, lipid A 4'-phosphatase activity prevents canonical bacterium-induced delay in antigen degradation, which leads to inefficient antigen cross-presentation and a failure to cross-prime CD8 T cells specific for a P. gingivalis-associated antigen. We propose that lipid A modifications produced by this bacterium alter host TLR4-dependent adaptive immunity to establish chronic infections associated with a number of systemic inflammatory disorders.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Apresentação Cruzada/fisiologia , Células Dendríticas/metabolismo , Imunidade Inata/fisiologia , Lipopolissacarídeos/metabolismo , Porphyromonas gingivalis/metabolismo , Porphyromonas gingivalis/patogenicidade , Variação Genética , Genótipo , Interações Hospedeiro-Patógeno , Humanos , Periodonto/microbiologia , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/imunologia
14.
BMC Complement Med Ther ; 20(1): 62, 2020 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-32106858

RESUMO

BACKGROUND: Natural products constitute a promising class of therapeutics for the treatment of gingivitis and periodontitis as well as the maintenance of oral health. However, the limited understanding behind their potential mechanisms and modes of action have hampered their incorporation into popular western therapeutics. This in vitro study characterizes an Ayurvedic herbal extract mixture, which has been clinically shown to promote gingival health and homeostasis. METHODS: Telomerase immortalized gingival keratinocytes (TIGK) were infected with either Fusobacterium nucleatum cell wall, live F. nucleatum, IL-1ß or TNF-α for 4 hours with and without the herbal extract. The immunomodulatory effects of the extract on host IL-8 production was measured by ELISA. RESULTS: It was found that the Ayurvedic herbal extract mixture inhibited gingival epithelial cell IL-8 expression in response to both bacterial and host cytokine agonists. The herbal extract inhibited IL-8 stimulated by F. nucleatum cell wall, live F. nucleatum, IL-1ß, and TNF-α in a dose-dependent manner that was not a result of host cell death. Furthermore, the extract showed significantly different ID50 doses demonstrating the differential ability to modulate both stimulated and basal IL-8 levels. CONCLUSIONS: In vitro investigation of this herbal extract mixture revealed that it has the ability to modulate gingival epithelial cell IL-8 expression in response to stimulation by bacterial components and host pro-inflammatory signals. This data demonstrates that the reduction in the gingival epithelial cell IL-8 response may in part be responsible for the previously reported ability of the Ayurvedic herbal extract mixture to reduce gingivitis in two separate human clinical studies.


Assuntos
Células Epiteliais/efeitos dos fármacos , Gengiva/efeitos dos fármacos , Interleucina-8/metabolismo , Ayurveda , Periodontite/tratamento farmacológico , Extratos Vegetais/farmacologia , Linhagem Celular , Fusobacterium nucleatum/efeitos dos fármacos , Gengiva/citologia , Humanos , Interleucina-1beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
15.
Artigo em Inglês | MEDLINE | ID: mdl-33604521

RESUMO

OBJECTIVE: Sudantha® (SUD), a natural proprietary mixture of herbal extracts that has been incorporated into toothpaste, has been shown in two separate placebo controlled human clinical studies to promote gingival health; and reduce gingival bleeding and plaque formation. However, the herbal based anti-gingivitis mechanisms of Sudantha are not fully understood. The objective of this study was to determine the effect of Sudantha on dental plaque biofilms by investigating its effect on mono-culture biofilms of a primary colonizer, Streptococcus mutans, in vitro. RESULTS: This study found that SUD contributes to the maintenance of oral health through the inhibition of S. mutans biofilm formation. In addition, SUD disrupted preformed S. mutans biofilms after exposure to SUD for 4 hours. Together, this pilot data suggests the inhibition of S. mutans biofilm formation and disruption represents one potential mechanism by which the herbal extract is able to reduce the oral bacterial biofilm resulting in its effective against gingivitis and its potential use in countering biofilm associated oral disease.

16.
Adv Exp Med Biol ; 1197: 55-67, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31732934

RESUMO

Gingival epithelium plays a pivotal role in protecting the underlying periodontium from the microbial colonization found in the gingival sulcus. Having an appropriate phenotype displayed by gingival epithelial cells is a critical host component required for protection against bacterial invasion into gingival tissues. In the present study, gingival epithelial homeostasis associated with the CXCL-8/IL-8 chemokine response was investigated in vitro to determine the mechanisms that gingival epithelial cells utilize for sensing gram-positive and gram-negative microorganisms. The findings of this study have demonstrated, by using Fusobacterium nucleatum, a heterogeneity of gingival epithelial cell response by Toll-like receptor (TLR) 2, a lipoprotein sensor. Notably, however, lipopolysaccharide (LPS), a major virulence factor of gram-negative bacteria, is not recognized by gingival epithelial cells unless the LPS is internalized into the cells. Activation of TLR4 in gingival epithelial cells occurs in the endosome, an intracellular event that requires a vesicular acidification to turn on TLR4 signaling, indicating their stringency for fine-tuning a local LPS response. This study has identified a unique LPS sensing mechanism of the oral epithelium to overcome a periodontal infection associated with LPS derived from gram-negative microbes that arises during dysbiosis.


Assuntos
Gengiva , Lipopolissacarídeos , Periodontite , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Gengiva/citologia , Gengiva/imunologia , Gengiva/microbiologia , Humanos , Interleucina-8/imunologia , Lipopolissacarídeos/metabolismo , Periodontite/imunologia , Periodontite/microbiologia
17.
Infect Immun ; 87(12)2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31570556

RESUMO

The Porphyromonas gingivalis strain ATCC 33277 (33277) and 381 genomes are nearly identical. However, strain 33277 displays a significantly diminished capacity to stimulate host cell Toll-like receptor 2 (TLR2)-dependent signaling and interleukin-1ß (IL-1ß) production relative to 381, suggesting that there are strain-specific differences in one or more bacterial immune-modulatory factors. Genomic sequencing identified a single nucleotide polymorphism in the 33277 fimB allele (A→T), creating a premature stop codon in the 33277 fimB open reading frame relative to the 381 fimB allele. Gene exchange experiments established that the 33277 fimB allele reduces the immune-stimulatory capacity of this strain. Transcriptome comparisons revealed that multiple genes related to carboxy-terminal domain (CTD) family proteins, including the gingipains, were upregulated in 33277 relative to 381. A gingipain substrate degradation assay demonstrated that cell surface gingipain activity is higher in 33277, and an isogenic mutant strain deficient for the gingipains exhibited an increased ability to induce TLR2 signaling and IL-1ß production. Furthermore, 33277 and 381 mutant strains lacking CTD cell surface proteins were more immune-stimulatory than the parental wild-type strains, consistent with an immune-suppressive role for the gingipains. Our data show that the combination of an intact fimB allele and limited cell surface gingipain activity in P. gingivalis 381 renders this strain more immune-stimulatory. Conversely, a defective fimB allele and high-level cell surface gingipain activity reduce the capacity of P. gingivalis 33277 to stimulate host cell innate immune responses. In summary, genomic and transcriptomic comparisons identified key virulence characteristics that confer divergent host cell innate immune responses to these highly related P. gingivalis strains.


Assuntos
Proteínas de Fímbrias/genética , Proteínas de Fímbrias/imunologia , Cisteína Endopeptidases Gingipaínas/metabolismo , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/imunologia , Infecções por Bacteroidaceae/imunologia , Infecções por Bacteroidaceae/microbiologia , Linhagem Celular Tumoral , Células HEK293 , Humanos , Imunidade Inata/genética , Imunidade Inata/imunologia , Interleucina-1beta/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Transdução de Sinais/imunologia , Células THP-1 , Receptor 2 Toll-Like/metabolismo
18.
J Periodontol ; 90(10): 1202-1212, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31111967

RESUMO

BACKGROUND: Oral gingival tissue, especially the junctional epithelium (JE), is constantly exposed to sub-gingival plaque. A key component of gingival health is the regulation of the number of neutrophils that migrate into the gingival crevice to counteract its harmful effects. This report investigates the contribution of innate defense receptors, Toll-like receptor (TLR)2, TLR4, and both (TLR2/4) to the maintenance of neutrophil homeostasis in the JE. METHODS: Bacterial composition was analyzed from whole oral swabs collected from 12- to 14-week-old TLR2, TLR4, TLR2/4 double knock-out (KO) mice using a MiSeq platform targeting the V3-V4 region of the 16S ribosomal RNA gene. Mandibles were histologically examined for quantification of neutrophils in the JE and bone loss. Lastly, total bacterial load was quantitated using quantitative real-time PCR. RESULTS: Compared with wild-type, all TLR KO mice displayed significantly increased recruitment of neutrophils (P = 0.0079) into the JE. In addition, TLR4 and TLR2/4 KO mice demonstrated a significant increase in the number of bacteria (P = 0.0022 and P = 0.0152, respectively). Lastly, comparative compositional analyses of the oral microbiome revealed that each KO strain harbored unique microbial communities that are distinct from each other but maintained similar levels of alveolar bone. CONCLUSIONS: Neutrophil migration into healthy mouse JE does not require TLR2 or TLR4. However, a significant increase in the number of neutrophils as well as a significant change in the oral microbial composition in both TLR2 and TLR4 KO mice demonstrate that these TLRs contribute to the homeostatic relationship between bacteria and the host in healthy mice periodontal tissue.


Assuntos
Microbiota , Receptor 2 Toll-Like , Animais , Inserção Epitelial , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos , Receptor 4 Toll-Like
19.
J Bacteriol ; 201(11)2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30782639

RESUMO

Removal of one acyl chain from bacterial lipid A by deacylase activity is a mechanism used by many pathogenic bacteria to evade the host's Toll-like receptor 4 (TLR4)-mediated innate immune response. In Porphyromonas gingivalis, a periodontal pathogen, lipid A deacylase activity converts a majority of the initially synthesized penta-acylated lipid A, a TLR4 agonist, to tetra-acylated structures, which effectively evade TLR4 sensing by being either inert or antagonistic at TLR4. In this paper, we report successful identification of the gene that encodes the P. gingivalis lipid A deacylase enzyme. This gene, PGN_1123 in P. gingivalis 33277, is highly conserved within P. gingivalis, and putative orthologs are phylogenetically restricted to the Bacteroidetes phylum. Lipid A of ΔPGN_1123 mutants is penta-acylated and devoid of tetra-acylated structures, and the mutant strain provokes a strong TLR4-mediated proinflammatory response, in contrast to the negligible response elicited by wild-type P. gingivalis Heterologous expression of PGN_1123 in Bacteroides thetaiotaomicron promoted lipid A deacylation, confirming that PGN_1123 encodes the lipid A deacylase enzyme.IMPORTANCE Periodontitis, commonly referred to as gum disease, is a chronic inflammatory condition that affects a large proportion of the population. Porphyromonas gingivalis is a bacterium closely associated with periodontitis, although how and if it is a cause for the disease are not known. It has a formidable capacity to dampen the host's innate immune response, enabling its persistence in diseased sites and triggering microbial dysbiosis in animal models of infection. P. gingivalis is particularly adept at evading the host's TLR4-mediated innate immune response by modifying the structure of lipid A, the TLR4 ligand. In this paper, we report identification of the gene encoding lipid A deacylase, a key enzyme that modifies lipid A to TLR4-evasive structures.


Assuntos
Proteínas de Bactérias/genética , Hidrolases de Éster Carboxílico/genética , Regulação Bacteriana da Expressão Gênica , Evasão da Resposta Imune/genética , Lipídeo A/química , Porphyromonas gingivalis/genética , Receptor 4 Toll-Like/genética , Carga Bacteriana , Proteínas de Bactérias/metabolismo , Bacteroides thetaiotaomicron/genética , Bacteroides thetaiotaomicron/metabolismo , Hidrolases de Éster Carboxílico/metabolismo , Linhagem Celular , Sequência Conservada , Células HEK293 , Humanos , Lipídeo A/imunologia , Monócitos/imunologia , Monócitos/microbiologia , Porphyromonas gingivalis/metabolismo , Receptor 4 Toll-Like/imunologia
20.
PLoS One ; 12(4): e0173223, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28384159

RESUMO

Lipopolysaccharide (LPS)-binding protein (LBP) as an acute-phase protein plays a crucial role in innate host response to bacterial challenge. Our previous study shows that LBP expression in human gingiva is associated with periodontal status. Porphyromonas gingivalis is a keystone periodontopathogen, and its LPS with lipid A structural heterogeneity critically accounts for periodontal pathogenesis. This study investigated the effects of LBP and its interactions with two featured isoforms of P. gingivalis LPS (tetra-acylated LPS1435/1449 and penta-acylated LPS1690) on the expression of pro-inflammatory cytokines in human oral keratinocytes (HOKs), and the involvement of Toll-like receptor (TLR) signaling. HOKs were pre-incubated with recombinant human LBP (rhLBP) at 10ng/ml, 100ng/ml and 1µg/ml for 1 h, followed by the treatment of P. gingivalis LPS1690 or LPS1435/1449 for 3h or 24h respectively. The expression of IL-6 and IL-8, and involvements of TLR2 and TLR4 were analyzed. The genes associated with TLR signaling were assessed by PCR array. Interestingly, rhLBP per se significantly up-regulated the expression of IL-6 and IL-8 in HOKs (p<0.05), which was blocked by TLR2 antibody (p<0.001). LPS1435/1449 down-regulated more significantly rhLBP-induced IL-6 and IL-8 mRNAs with reference to P. gingivalis LPS1690 (approximately 80% vs. 40%, p<0.05; and 90% vs. 36%, p<0.001, respectively). Moreover, rhLBP markedly down-regulated the gene expression of TLRs and their adaptors such as CD180 (-2.44 folds) and MD-1 (-9.62 folds), while the interaction of P. gingivalis LPS1435/1449 with rhLBP greatly up-regulated both transcripts (7.11 and 4.05 folds, respectively). Notably, P. gingivalis LPS1690-rhLBP interaction dramatically up-regulated CD180 transcript (20.86 folds) and significantly down-regulated MD-1 transcript (-6.93 folds). This pioneering study shows that rhLBP enables to enhance the expression of pro-inflammatory cytokines in HOKs through TLR2 signaling pathway. P. gingivalis LPS with different lipid A structures down-regulates to different extents rhLBP-induced cytokine expression, possibly through fine-tuning of the CD180-MD1 complex and relevant TLRs.


Assuntos
Proteínas de Fase Aguda/metabolismo , Proteínas de Transporte/metabolismo , Inflamação/metabolismo , Queratinócitos/metabolismo , Lipopolissacarídeos/metabolismo , Glicoproteínas de Membrana/metabolismo , Porphyromonas gingivalis/metabolismo , Transdução de Sinais , Receptores Toll-Like/metabolismo , Células Cultivadas , Humanos , Queratinócitos/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...